bims-imseme Biomed News
on Immunosenescence and T cell metabolism
Issue of 2020‒08‒16
nineteen papers selected by
Pierpaolo Ginefra
Ludwig Institute for Cancer Research


  1. Cell Metab. 2020 Jul 31. pii: S1550-4131(20)30366-1. [Epub ahead of print]
      Dysregulated metabolism is a key driver of maladaptive tumor-reactive T lymphocytes within the tumor microenvironment. Actionable targets that rescue the effector activity of antitumor T cells remain elusive. Here, we report that the Sirtuin-2 (Sirt2) NAD+-dependent deacetylase inhibits T cell metabolism and impairs T cell effector functions. Remarkably, upregulation of Sirt2 in human tumor-infiltrating lymphocytes (TILs) negatively correlates with response to TIL therapy in advanced non-small-cell lung cancer. Mechanistically, Sirt2 suppresses T cell metabolism by targeting key enzymes involved in glycolysis, tricarboxylic acid-cycle, fatty acid oxidation, and glutaminolysis. Accordingly, Sirt2-deficient murine T cells exhibit increased glycolysis and oxidative phosphorylation, resulting in enhanced proliferation and effector functions and subsequently exhibiting superior antitumor activity. Importantly, pharmacologic inhibition of Sirt2 endows human TILs with these superior metabolic fitness and effector functions. Our findings unveil Sirt2 as an unexpected actionable target for reprogramming T cell metabolism to augment a broad spectrum of cancer immunotherapies.
    Keywords:  FAO; OxPhos; Sirt2; T cells; antitumor immunity; deacetylase; dysregulated metabolism; glutaminolysis; glycolysis; metabolic checkpoint
    DOI:  https://doi.org/10.1016/j.cmet.2020.07.008
  2. Dev Comp Immunol. 2020 Aug 11. pii: S0145-305X(20)30364-5. [Epub ahead of print] 103809
      Goats are important food animals and are disseminated globally because of their high adaptability to varying environmental conditions and feeding regimes that provide them with a comparative advantage. Productivity is impacted by infectious diseases; this then contributes to societal poverty, food insecurity, and international trade restrictions. Since γδ T cells have been shown to have vital roles in immune responses in other mammals we reviewed the literature regarding what is known about their functions, distribution in tissues and organs and their responses to a variety of infections in goats. It has been shown that caprine γδ T cells produce interferon-γ and IL-17, are found in a variety of lymphoid and nonlymphoid tissues and constitute a significant population of blood mononuclear cells. Their representation in tissues and their functional responses may be altered concomitant with infection. This review summarizes caprine γδ T cell responses to Brucella melitensis, Fasciola hepatica, Mycobacterium avium paratuberculosis, caprine arthritis encephalitis virus (CAEV), and Schistosoma bovis in infected or vaccinated goats. Caprine γδ T cells have also been evaluated in goats infected with M. caprae, Ehrilichia ruminantium, Haemonchus contortus and peste des petits ruminants (PPR) virus but found to have an unknown or limited response or role in either protective immunity or immunopathogenesis in those cases.
    Keywords:  Goat; Immunity; Infectious diseases; T cells; T lymphocytes; γδ T cells
    DOI:  https://doi.org/10.1016/j.dci.2020.103809
  3. Front Immunol. 2020 ;11 1517
      Allogeneic hematopoietic stem cell transplantation (aHSCT) is a curative therapy for a range of hematologic illnesses including aplastic anemia, sickle cell disease, immunodeficiency, and high-risk leukemia, but the efficacy of aHSCT is often undermined by graft-versus-host disease (GVHD), where T cells from the donor attack and destroy recipient tissues. Given the strong interconnection between T cell metabolism and cellular function, determining the metabolic pathways utilized by alloreactive T cells is fundamental to deepening our understanding of GVHD biology, including its initiation, propagation, and potential mitigation. This review summarizes the metabolic pathways available to alloreactive T cells and highlights key metabolic proteins and pathways linking T cell metabolism to effector function. Our current knowledge of alloreactive T cell metabolism is then explored, showing support for glycolysis, fat oxidation, and glutamine metabolism but also offering a potential explanation for how these presumably contradictory metabolic findings might be reconciled. Examples of additional ways in which metabolism impacts aHSCT are addressed, including the influence of butyrate metabolism on GVHD resolution. Finally, the caveats and challenges of assigning causality using our current metabolic toolbox is discussed, as well as likely future directions in immunometabolism, both to highlight the strengths of the current evidence as well as recognize some of its limitations.
    Keywords:  AMPK; GVHD biology; alloreactive T cells; fatty acid oxidation (FAO); glycolysis; immunometabolism; mammalian target of rapamycin (mTOR)
    DOI:  https://doi.org/10.3389/fimmu.2020.01517
  4. Semin Immunopathol. 2020 Aug 12.
      Obesity represents a serious health problem as it is rapidly increasing worldwide. Obesity is associated with reduced healthspan and lifespan, decreased responses to infections and vaccination, and increased frequency of inflammatory conditions typical of old age. Obesity is characterized by increased fat mass and remodeling of the adipose tissue (AT). In this review, we summarize published data on the different types of AT present in mice and humans, and their roles as fat storage as well as endocrine and immune tissues. We review the age-induced changes, including those in the distribution of fat in the body, in abundance and function of adipocytes and their precursors, and in the infiltration of immune cells from the peripheral blood. We also show that cells with a senescent-associated secretory phenotype accumulate in the AT of mice and humans with age, where they secrete several factors involved in the establishment and maintenance of local inflammation, oxidative stress, cell death, tissue remodeling, and infiltration of pro-inflammatory immune cells. Not only adipocytes and pre-adipocytes but also immune cells show a senescent phenotype in the AT. With the increase in human lifespan, it is crucial to identify strategies of intervention and target senescent cells in the AT to reduce local and systemic inflammation and the development of age-associated diseases. Several studies have indeed shown that senescent cells can be effectively targeted in the AT by selectively removing them or by inhibiting the pathways that lead to the secretion of pro-inflammatory factors.
    Keywords:  Adipocytes; Adipose tissue; Aging; Cell senescence; Immune cells
    DOI:  https://doi.org/10.1007/s00281-020-00812-1
  5. Blood. 2020 Aug 12. pii: blood.2020005770. [Epub ahead of print]
      Graft-versus-host disease (GvHD) is a prominent barrier to allogeneic hematopoietic stem cell transplantation (AHSCT). Definitive diagnosis of GvHD is invasive and biopsies of involved tissues pose a high risk of bleeding and infection. T cells are central to GvHD pathogenesis and our previous studies in a chronic GvHD mouse model showed that alloreactive CD4+ T cells traffic to the target organs ahead of overt symptoms. Since increased glycolysis is an early feature of T cell activation, we hypothesized that in vivo metabolic imaging of glycolysis would allow non-invasive detection of liver GvHD as activated CD4+ T cells traffic into the organ. Indeed, hyperpolarized 13C-pyruvate MRI detected high rates of conversion of pyruvate to lactate in the liver ahead of animals becoming symptomatic, but not during subsequent overt cGvHD. Concomitantly, CD4+ T effector memory cells, the predominant pathogenic CD4+ T cell subset, were confirmed to be highly glycolytic by transcriptomic, protein, metabolite, and ex vivo metabolic activity analyses. Preliminary data from single cell sequencing of circulating T cells in patients undergoing allogeneic HSCT also suggested that increased glycolysis may be a feature of incipient acute GvHD. Metabolic imaging is being increasingly used in the clinic and may be useful in the post-AHSCT setting for non-invasive early detection of GvHD.
    DOI:  https://doi.org/10.1182/blood.2020005770
  6. Inflamm Res. 2020 Aug 14.
      PURPOSE: Rheumatoid arthritis (RA) is a chronic, systemic autoimmune disease with synovitis as pathological changes. The immune microenvironment of RA promotes metabolic reprogramming of immune cells and stromal cells, which leads to dysfunction and imbalance of immune homeostasis. Cell metabolism undergoes the switch from a static regulatory state to a highly metabolic active state, which changes the redox-sensitive signaling pathway and also leads to the accumulation of metabolic intermediates, which in turn can act as signaling molecules and further aggravate the inflammatory response. The reprogramming of immunometabolism affects the function of immune cells and is crucial to the pathogenesis of RA. In addition, mitochondrial dysfunction plays a key role in glycolytic reprogramming in RA. These metabolic changes may be potential therapeutic targets for RA. Therefore, we reviewed the metabolic reprogramming of RA immune cells and fibroblast-like synovium cells (FLS) and its relationship with mitochondrial dysfunction.METHODS: A computer-based online search was performed using the PubMed database and Web of Science database for published articles concerning immunometabolic reprogramming, mitochondrial dysfunction, and rheumatoid arthritis.
    RESULTS: This article reviews the metabolic reprogramming of immune cells and fibroblast-like synoviocytes in RA and their relationship to mitochondrial disfunction, as well as the key pro-inflammatory pathways associated with metabolic reprogramming and chemotherapy as a potential future therapeutic strategy for RA.
    Keywords:  Fibroblast-like synoviocytes; Macrophages; Metabolic reprogramming; Mitochondrial dysfunction; Rheumatoid arthritis; T cells
    DOI:  https://doi.org/10.1007/s00011-020-01391-5
  7. Metabolism. 2020 Aug 10. pii: S0026-0495(20)30202-X. [Epub ahead of print] 154338
      Peroxisome proliferator-activated receptors (PPARs) are fatty acid-activated transcription factors of nuclear hormone receptor superfamily that regulate energy metabolism. Currently, three PPAR subtypes have been identified: PPARα, PPARγ, and PPARβ/δ. PPARα and PPARδ are highly expressed in oxidative tissues and regulate genes involved in substrate delivery and oxidative phosphorylation (OXPHOS) and regulation of energy homeostasis. In contrast, PPARγ is more important in lipogenesis and lipid synthesis, with highest expression levels in white adipose tissue (WAT). In addition to tissues regulating whole body energy homeostasis, PPARs are expressed in immune cells and have an emerging critical role in immune cell differentiation and fate commitment. In this review, we discuss the actions of PPARs in the function of the innate and the adaptive immune system and their implications in immune-mediated inflammatory conditions.
    Keywords:  PPAR; T cells; inflammation; metabolism; myeloid cells
    DOI:  https://doi.org/10.1016/j.metabol.2020.154338
  8. Nat Commun. 2020 Aug 07. 11(1): 3946
      Melanomas can switch to a dedifferentiated cell state upon exposure to cytotoxic T cells. However, it is unclear whether such tumor cells pre-exist in patients and whether they can be resensitized to immunotherapy. Here, we chronically expose (patient-derived) melanoma cell lines to differentiation antigen-specific cytotoxic T cells and observe strong enrichment of a pre-existing NGFRhi population. These fractions are refractory also to T cells recognizing non-differentiation antigens, as well as to BRAF + MEK inhibitors. NGFRhi cells induce the neurotrophic factor BDNF, which contributes to T cell resistance, as does NGFR. In melanoma patients, a tumor-intrinsic NGFR signature predicts anti-PD-1 therapy resistance, and NGFRhi tumor fractions are associated with immune exclusion. Lastly, pharmacologic NGFR inhibition restores tumor sensitivity to T cell attack in vitro and in melanoma xenografts. These findings demonstrate the existence of a stable and pre-existing NGFRhi multitherapy-refractory melanoma subpopulation, which ought to be eliminated to revert intrinsic resistance to immunotherapeutic intervention.
    DOI:  https://doi.org/10.1038/s41467-020-17739-8
  9. Nat Rev Immunol. 2020 Aug 12.
      IL-9-producing CD4+ T cells have been considered to represent a distinct T helper cell (TH cell) subset owing to their unique developmental programme in vitro, their expression of distinct transcription factors (including PU.1) and their copious production of IL-9. It remains debatable whether these cells represent a truly unique TH cell subset in vivo, but they are closely related to the T helper 2 (TH2) cells that are detected in allergic diseases. In recent years, increasing evidence has also indicated that IL-9-producing T cells may have potent abilities in eradicating advanced tumours, particularly melanomas. Here, we review the latest literature on the development of IL-9-producing T cells and their functions in disease settings, with a particular focus on allergy and cancer. We also discuss recent ideas concerning the therapeutic targeting of these cells in patients with chronic allergic diseases and their potential use in cancer immunotherapy.
    DOI:  https://doi.org/10.1038/s41577-020-0396-0
  10. J Med Virol. 2020 Aug 13.
      Nutrient starvation is a common phenomenon that occurs during T cell activation. Upon pathogen infection, large amounts of immune cells migrate to infection sites, and antigen-specific T cells are activated; this is followed by rapid proliferation through clonal expansion. The dramatic expansion of cells will commonly lead to nutrient shortage. Cellular autophagy is often upregulated as a way to sustain the body's energy requirements. During infection, HIV co-opts a series of host cell metabolic pathways for replication. Several HIV proteins, such as Env, Nef, and Vpr, have already been reported as being involved in autophagy-related processes. In this report, we identified that the HIV p17 protein acts as a major factor in suppressing the autophagic process in T cells, especially under glucose starvation condition. HIV p17 interacts with OLA1 and disrupts OLA1-GSK3β complex, leading to GSK3β hyperactivation. Consequently, a prior proliferation of HIV-infected T cells under glucose starvation will occur. The inhibition of autophagy also aids HIV replication by antagonizing the antiviral effect of autophagy. Our study shows a new cellular pathway that HIV can hijack for viral spreading by a prior proliferation of HIV-loaded T cells and may provide new therapeutic targets for AIDS intervention. This article is protected by copyright. All rights reserved.
    Keywords:  HIV; T cell survival; autophagy; protein interaction; viral infection
    DOI:  https://doi.org/10.1002/jmv.26423
  11. Curr Osteoporos Rep. 2020 Aug 13.
      PURPOSE OF REVIEW: Although many signalling pathways have been discovered to be essential in mesenchymal stem/stromal (MSC) differentiation, it has become increasingly clear in recent years that epigenetic regulation of gene transcription is a vital component of lineage determination, encompassing diet, lifestyle and parental influences on bone, fat and cartilage development.RECENT FINDINGS: This review discusses how specific enzymes that modify histone methylation and acetylation or DNA methylation orchestrate the differentiation programs in lineage determination of MSC and the epigenetic changes that facilitate development of bone related diseases such as osteoporosis. The review also describes how environmental factors such as mechanical loading influence the epigenetic signatures of MSC, and how the use of chemical agents or small peptides can regulate epigenetic drift in MSC populations during ageing and disease. Epigenetic regulation of MSC lineage commitment is controlled through changes in enzyme activity, which modifies DNA and histone residues leading to alterations in chromatin structure. The co-ordinated epigenetic regulation of transcriptional activation and repression act to mediate skeletal tissue homeostasis, where deregulation of this process can lead to bone loss during ageing or osteoporosis.
    Keywords:  DNA methylation; Epigenetics; Histone acetylation; Histone methylation; MSC; Mesenchymal stem/stromal cells; Skeletal stem cells
    DOI:  https://doi.org/10.1007/s11914-020-00616-0
  12. Curr Opin Oncol. 2020 Sep;32(5): 398-407
      PURPOSE OF REVIEW: A number of clinical trials are currently testing chimeric antigen receptor (CAR) and T cell receptor (TCR) engineered T cells for the treatment of haematologic malignancies and selected solid tumours, and CD19-CAR-T cells have produced impressive clinical responses in B-cell malignancies. Here, we summarize the current state of the field, highlighting the key aspects required for the optimal application of CAR and TCR-engineered T cells for cancer immunotherapy.RECENT FINDINGS: Toxicities, treatment failure and disease recurrence have been observed at different rates and kinetics. Several strategies have been designed to overcome these hurdles: the identification and combination of known and new antigens, together with the combination of immunotherapeutic and classical approaches may overcome cancer immune evasion. New protocols for genetic modification and T cell culture may improve the overall fitness of cellular products and their resistance to hostile tumour immunomodulatory signals. Finally, the schedules of T cell administration and toxicity management have been adapted to improve the safety of this transformative therapeutic approach.
    SUMMARY: In order to develop effective adoptive T cell treatments for cancer, therapeutic optimization of engineered CAR and TCR T cells is crucial, by simultaneously focusing on intrinsic and extrinsic factors. This review focuses on the innovative approaches designed and tested to overcome the hurdles encountered so far in the clinical practice, with new excitement on novel laboratory insights and ongoing clinical investigations.
    DOI:  https://doi.org/10.1097/CCO.0000000000000664
  13. Front Oncol. 2020 ;10 1197
      Immune checkpoint inhibitors (ICIs) targeting immune checkpoint proteins, such as CTLA-4 and PD-1/PD-L1, have demonstrated remarkable and durable clinical responses in various cancer types. However, a considerable number of patients receiving ICIs eventually experience a relapse due to diverse resistance mechanisms. As a result, there have been increasing research efforts to elucidate the molecular mechanisms behind resistance to ICIs and improve patient outcomes. There is growing evidence that the dysregulated metabolic activity of tumor cells generates an immunosuppressive tumor microenvironment (TME) that orchestrates an impaired anti-tumor immune response. Notably, the immunosuppressive TME is characterized by nutrient shortage, hypoxia, an acidic extracellular milieu, and abundant immunosuppressive molecules. A detailed understanding of the TME remains a major challenge in mounting a more effective anti-tumor immune response. Herein, we discuss how tumor cells reprogram metabolism to modulate a pro-tumor TME, driving disease progression and immune evasion; in particular, we highlight potential approaches to target metabolic vulnerabilities in the context of anti-tumor immunotherapy.
    Keywords:  cancer cell metabolite; cancer metabolism; immune checkpoint inhibitors; immune evasion; tumor microenvironment
    DOI:  https://doi.org/10.3389/fonc.2020.01197
  14. Mol Biol Rep. 2020 Aug 12.
      S-acylation reversible-post-translational lipidation of cysteine residues-is emerging as an important regulatory mechanism in T cell signaling. Dynamic S-acylation is critical for protein recruitment into the T cell receptor complex and initiation of the subsequent signaling cascade. However, the enzymatic control of protein S-acylation in T cells remains poorly understood. Here, we report a previously uncharacterized role of DHHC21, a member of the mammalian family of DHHC protein acyltransferases, in regulation of the T cell receptor pathway. We found that loss of DHHC21 prevented S-acylation of key T cell signaling proteins, resulting in disruption of the early signaling events and suppressed expression of T cell activation markers. Furthermore, downregulation of DHHC21 prevented activation and differentiation of naïve T cells into effector subtypes. Together, our study provides the first direct evidence that DHHC protein acyltransferases can play an essential role in regulation of T cell-mediated immunity.
    Keywords:  Acyltransferase; Cell signaling; DHHC21; Protein acylation; Protein palmitoylation; Signal transduction; T helper cells; T-cell
    DOI:  https://doi.org/10.1007/s11033-020-05691-1
  15. Curr Opin Immunol. 2020 Aug 08. pii: S0952-7915(20)30071-6. [Epub ahead of print]67 10-17
      Over the last few years, immune cell metabolism has become one of the most stimulating areas of investigation in the field of immunology. Compelling evidence has revealed that metabolic pathways are closely associated to cell functions and immune cells adopt defined metabolic programs to sustain their activity and respond to micro-environmental demands. It is now clear that alterations in cell metabolism can favour dysregulation typical of autoreactive immune cells, thus sustaining loss of immunological self-tolerance. In this short review, we highlight the main metabolic alterations associated with both innate and adaptive immune cells in autoimmune conditions, such as multiple sclerosis (MS) and type 1 diabetes (T1D). We also summarize recent findings reporting the use of pharmacological agents, which modulate the immunometabolism to possibly control immune responses during autoimmune disorders.
    DOI:  https://doi.org/10.1016/j.coi.2020.07.002
  16. Immunometabolism. 2020 ;2(3): e200026
      Aging is a complex process that involves dysfunction on multiple levels, all of which seem to converge on inflammation. Macrophages are intimately involved in initiating and resolving inflammation, and their dysregulation with age is a primary contributor to inflammaging-a state of chronic, low-grade inflammation that develops during aging. Among the age-related changes that occur to macrophages are a heightened state of basal inflammation and diminished or hyperactive inflammatory responses, which seem to be driven by metabolic-dependent epigenetic changes. In this review article we provide a brief overview of mitochondrial functions and age-related changes that occur to macrophages, with an emphasis on how the inflammaging environment, senescence, and NAD decline can affect their metabolism, promote dysregulation, and contribute to inflammaging and age-related pathologies.
    Keywords:  CD38; NAD; SASP; aging; immunometabolism; inflammaging; macrophage; mitochondria; monocyte; senescence
    DOI:  https://doi.org/10.20900/immunometab20200026
  17. Cell Metab. 2020 Aug 04. pii: S1550-4131(20)30411-3. [Epub ahead of print]
      The Krebs cycle-derived metabolite itaconate is highly upregulated in inflammatory macrophages and exerts immunomodulatory effects through cysteine modifications on target proteins. The NLRP3 inflammasome, which cleaves IL-1β, IL-18, and gasdermin D, must be tightly regulated to avoid excessive inflammation. Here we provide evidence that itaconate modifies NLRP3 and inhibits inflammasome activation. Itaconate and its derivative, 4-octyl itaconate (4-OI), inhibited NLRP3 inflammasome activation, but not AIM2 or NLRC4. Conversely, NLRP3 activation was increased in itaconate-depleted Irg1-/- macrophages. 4-OI inhibited the interaction between NLRP3 and NEK7, a key step in the activation process, and "dicarboxypropylated" C548 on NLRP3. Furthermore, 4-OI inhibited NLRP3-dependent IL-1β release from PBMCs isolated from cryopyrin-associated periodic syndrome (CAPS) patients, and reduced inflammation in an in vivo model of urate-induced peritonitis. Our results identify itaconate as an endogenous metabolic regulator of the NLRP3 inflammasome and describe a process that may be exploited therapeutically to alleviate inflammation in NLRP3-driven disorders.
    Keywords:  IL-1β; NEK7; NLRP3; cysteine modification; immunometabolism; inflammasome; itaconate; macrophage; metabolite; pyroptosis
    DOI:  https://doi.org/10.1016/j.cmet.2020.07.016
  18. Acta Biochim Biophys Sin (Shanghai). 2020 Aug 12. pii: gmaa081. [Epub ahead of print]
      As a major type of immune cells with heterogeneity and plasticity, macrophages are classically divided into inflammatory (M1) and alternative/anti-inflammatory (M2) types and play a crucial role in the progress of the inflammatory diseases. Recent studies have shown that metabolism is an important determinant of macrophage phenotype. Mitochondria, one of the most important compartments involving cell metabolism, are closely associated with the regulation of cell functions. In most types of cell, mitochondrial oxidative phosphorylation (OXPHOS) is the primary mode of cellular energy production. However, mitochondrial OXPHOS is inhibited in activated M1 macrophages, rendering them unable to be converted into M2 phenotype. Thus, mitochondrial metabolism is a crucial regulator in macrophage functions. This review summarizes the roles of mitochondria in macrophage polarization and analyzes the molecular mechanisms underlying mitochondrial metabolism and function, which may provide new approaches for the treatment of metabolic inflammatory diseases.
    Keywords:  inflammation; macrophage polarization; metabolism; mitochondria
    DOI:  https://doi.org/10.1093/abbs/gmaa081